36,605 results on '"Catalytic Domain"'
Search Results
2. In Crystallo O2 Cleavage at a Preorganized Triiron Cluster
- Author
-
Lee, Heui Beom, Ciolkowski, Nicholas, Field, Mackenzie, Marchiori, David A, Britt, R David, Green, Michael T, and Rittle, Jonathan
- Subjects
Inorganic Chemistry ,Chemical Sciences ,Oxygen ,Iron ,Crystallography ,X-Ray ,Catalytic Domain ,Oxidation-Reduction ,Models ,Molecular ,General Chemistry ,Chemical sciences ,Engineering - Abstract
In Nature, the four-electron reduction of O2 is catalyzed at preorganized multimetallic active sites. These complex active sites often feature low-coordinate, redox-active metal centers precisely positioned to facilitate rapid O2 activation processes that obviate the generation of toxic, partially reduced oxygen species. Very few biomimetic constructs simultaneously recapitulate the complexity and reactivity of these biological cofactors. Herein, we report solid-state O2 activation at a triiron(II) active site templated by phosphinimide ligands. Insight into the structure of the O2 reduction intermediates was obtained via in crystallo O2 dosing experiments in conjunction with spectroscopic, structural, magnetic, and computational studies. These data support the in situ formation of an Fe2IIIFeIV-dioxo intermediate upon exposure to O2 that participates in oxygen atom and hydrogen atom transfer reactivity with exogenous substrates to furnish a stable FeIIFe2III-oxo species. Combined, these studies provide an extraordinary level of detail into the dynamics of bond-forming and -breaking processes operative at complex multimetallic active sites.
- Published
- 2025
3. Carbohydrate Deacetylase Unique to Gut Microbe Bacteroides Reveals Atypical Structure
- Author
-
Schwartz, Lilith A, Norman, Jordan O, Hasan, Sharika, Adamek, Olive E, Dzuong, Elisa, Lowenstein, Jasmine C, Yost, Olivia G, Sankaran, Banumathi, and McLaughlin, Krystle J
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Autoimmune Disease ,Digestive Diseases ,Microbiome ,2.1 Biological and endogenous factors ,1.1 Normal biological development and functioning ,Oral and gastrointestinal ,Bacteroides ,Crystallography ,X-Ray ,Gastrointestinal Microbiome ,Amidohydrolases ,Humans ,Models ,Molecular ,Bacterial Proteins ,Catalytic Domain ,Protein Conformation ,Medicinal and Biomolecular Chemistry ,Medical Biochemistry and Metabolomics ,Biochemistry & Molecular Biology ,Biochemistry and cell biology ,Medical biochemistry and metabolomics ,Medicinal and biomolecular chemistry - Abstract
Bacteroides are often the most abundant, commensal species in the gut microbiome of industrialized human populations. One of the most commonly detected species is Bacteroides ovatus. It has been linked to benefits like the suppression of intestinal inflammation but is also correlated with some autoimmune disorders, for example irritable bowel disorder (IBD). Bacterial cell surface carbohydrates, like capsular polysaccharides (CPS), may play a role in modulating these varied host interactions. Recent studies have begun to explore the diversity of CPS loci in Bacteroides; however, there is still much unknown. Here, we present structural and functional characterization of a putative polysaccharide deacetylase from Bacteroides ovatus (BoPDA) encoded in a CPS biosynthetic locus. We solved four high resolution crystal structures (1.36-1.56 Å) of the enzyme bound to divalent cations Co2+, Ni2+, Cu2+, or Zn2+ and performed carbohydrate binding and deacetylase activity assays. Structural analysis of BoPDA revealed an atypical domain architecture that is unique to this enzyme, with a carbohydrate esterase 4 (CE4) superfamily catalytic domain inserted into a carbohydrate binding module (CBM). Additionally, BoPDA lacks the canonical CE4 His-His-Asp metal binding motif and our structures show it utilizes a noncanonical His-Asp dyad to bind metal ions. BoPDA is the first protein involved in CPS biosynthesis from B. ovatus to be characterized, furthering our understanding of significant biosynthetic processes in this medically relevant gut microbe.
- Published
- 2025
4. Coupling sensor to enzyme in the voltage sensing phosphatase.
- Author
-
Yu, Yawei, Zhang, Lin, Li, Baobin, Fu, Zhu, Brohawn, Stephen, and Isacoff, Ehud
- Subjects
Phosphoric Monoester Hydrolases ,Catalytic Domain ,Animals ,Protein Domains ,Models ,Molecular ,Mutation ,Humans ,Patch-Clamp Techniques - Abstract
Voltage-sensing phosphatases (VSPs) dephosphorylate phosphoinositide (PIP) signaling lipids in response to membrane depolarization. VSPs possess an S4-containing voltage sensor domain (VSD), resembling that of voltage-gated cation channels, and a lipid phosphatase domain (PD). The mechanism by which voltage turns on enzyme activity is unclear. Structural analysis and modeling suggest several sites of VSD-PD interaction that could couple voltage sensing to catalysis. Voltage clamp fluorometry reveals voltage-driven rearrangements in three sites implicated earlier in enzyme activation-the VSD-PD linker, gating loop and R loop-as well as the N-terminal domain, which has not yet been explored. N-terminus mutations perturb both rearrangements in the other segments and enzyme activity. Our results provide a model for a dynamic assembly by which S4 controls the catalytic site.
- Published
- 2024
5. A eukaryotic-like ubiquitination system in bacterial antiviral defence.
- Author
-
Chambers, Lydia, Ye, Qiaozhen, Cai, Jiaxi, Gong, Minheng, Ledvina, Hannah, Zhou, Huilin, Whiteley, Aaron, Suhandynata, Raymond, and Corbett, Kevin
- Subjects
Bacterial Proteins ,Bacteriophages ,Catalytic Domain ,Crystallography ,X-Ray ,Cysteine ,Deubiquitinating Enzymes ,Escherichia ,Evolution ,Molecular ,Lysine ,Models ,Molecular ,Operon ,Protein Domains ,Ubiquitin-Activating Enzymes ,Ubiquitin-Conjugating Enzymes ,Ubiquitination ,Ubiquitins ,Eukaryota - Abstract
Ubiquitination pathways have crucial roles in protein homeostasis, signalling and innate immunity1-3. In these pathways, an enzymatic cascade of E1, E2 and E3 proteins conjugates ubiquitin or a ubiquitin-like protein (Ubl) to target-protein lysine residues4. Bacteria encode ancient relatives of E1 and Ubl proteins involved in sulfur metabolism5,6, but these proteins do not mediate Ubl-target conjugation, leaving open the question of whether bacteria can perform ubiquitination-like protein conjugation. Here we demonstrate that a bacterial operon associated with phage defence islands encodes a complete ubiquitination pathway. Two structures of a bacterial E1-E2-Ubl complex reveal striking architectural parallels with canonical eukaryotic ubiquitination machinery. The bacterial E1 possesses an amino-terminal inactive adenylation domain and a carboxy-terminal active adenylation domain with a mobile α-helical insertion containing the catalytic cysteine (CYS domain). One structure reveals a pre-reaction state with the bacterial Ubl C terminus positioned for adenylation, and a second structure mimics an E1-to-E2 transthioesterification state with the E1 CYS domain adjacent to the bound E2. We show that a deubiquitinase in the same pathway preprocesses the bacterial Ubl, exposing its C-terminal glycine for adenylation. Finally, we show that the bacterial E1 and E2 collaborate to conjugate Ubl to target-protein lysine residues. Together, these data reveal that bacteria possess bona fide ubiquitination systems with strong mechanistic and architectural parallels to canonical eukaryotic ubiquitination pathways, suggesting that these pathways arose first in bacteria.
- Published
- 2024
6. Mutation-induced shift of the photosystem II active site reveals insight into conserved water channels.
- Author
-
Flesher, David, Liu, Jinchan, Wang, Jimin, Gisriel, Christopher, Yang, Ke, Batista, Victor, Debus, Richard, and Brudvig, Gary
- Subjects
cryo-EM ,hydrogen-bond network ,mutagenesis ,oxygen-evolving complex ,photosynthesis ,photosystem II ,quantum mechanics/molecular mechanics ,water channel ,Photosystem II Protein Complex ,Water ,Catalytic Domain ,Oxidation-Reduction ,Mutation ,Cryoelectron Microscopy ,Manganese - Abstract
Photosystem II (PSII) is the water-plastoquinone photo-oxidoreductase central to oxygenic photosynthesis. PSII has been extensively studied for its ability to catalyze light-driven water oxidation at a Mn4CaO5 cluster called the oxygen-evolving complex (OEC). Despite these efforts, the complete reaction mechanism for water oxidation by PSII is still heavily debated. Previous mutagenesis studies have investigated the roles of conserved amino acids, but these studies have lacked a direct structural basis that would allow for a more meaningful interpretation. Here, we report a 2.14-Å resolution cryo-EM structure of a PSII complex containing the substitution Asp170Glu on the D1 subunit. This mutation directly perturbs a bridging carboxylate ligand of the OEC, which alters the spectroscopic properties of the OEC without fully abolishing water oxidation. The structure reveals that the mutation shifts the position of the OEC within the active site without markedly distorting the Mn4CaO5 cluster metal-metal geometry, instead shifting the OEC as a rigid body. This shift disturbs the hydrogen-bonding network of structured waters near the OEC, causing disorder in the conserved water channels. This mutation-induced disorder appears consistent with previous FTIR spectroscopic data. We further show using quantum mechanics/molecular mechanics methods that the mutation-induced structural changes can affect the magnetic properties of the OEC by altering the axes of the Jahn-Teller distortion of the Mn(III) ion coordinated to D1-170. These results offer new perspectives on the conserved water channels, the rigid body property of the OEC, and the role of D1-Asp170 in the enzymatic water oxidation mechanism.
- Published
- 2024
7. Multisubstrate specificity shaped the complex evolution of the aminotransferase family across the tree of life
- Author
-
Koper, Kaan, Han, Sang-Woo, Kothadia, Ramani, Salamon, Hugh, Yoshikuni, Yasuo, and Maeda, Hiroshi A
- Subjects
Biochemistry and Cell Biology ,Evolutionary Biology ,Biological Sciences ,Generic health relevance ,Substrate Specificity ,Transaminases ,Evolution ,Molecular ,Phylogeny ,Catalytic Domain ,Nitrogen ,enzyme family evolution ,core metabolism ,substrate promiscuity ,nitrogen metabolism ,multifunctional enzymes - Abstract
Aminotransferases (ATs) are an ancient enzyme family that play central roles in core nitrogen metabolism, essential to all organisms. However, many of the AT enzyme functions remain poorly defined, limiting our fundamental understanding of the nitrogen metabolic networks that exist in different organisms. Here, we traced the deep evolutionary history of the AT family by analyzing AT enzymes from 90 species spanning the tree of life (ToL). We found that each organism has maintained a relatively small and constant number of ATs. Mapping the distribution of ATs across the ToL uncovered that many essential AT reactions are carried out by taxon-specific AT enzymes due to wide-spread nonorthologous gene displacements. This complex evolutionary history explains the difficulty of homology-based AT functional prediction. Biochemical characterization of diverse aromatic ATs further revealed their broad substrate specificity, unlike other core metabolic enzymes that evolved to catalyze specific reactions today. Interestingly, however, we found that these AT enzymes that diverged over billion years share common signatures of multisubstrate specificity by employing different nonconserved active site residues. These findings illustrate that AT family enzymes had leveraged their inherent substrate promiscuity to maintain a small yet distinct set of multifunctional AT enzymes in different taxa. This evolutionary history of versatile ATs likely contributed to the establishment of robust and diverse nitrogen metabolic networks that exist throughout the ToL. The study provides a critical foundation to systematically determine diverse AT functions and underlying nitrogen metabolic networks across the ToL.
- Published
- 2024
8. Structural and mechanistic insights into a lysosomal membrane enzyme HGSNAT involved in Sanfilippo syndrome.
- Author
-
Zhao, Boyang, Cao, Zhongzheng, Zheng, Yi, Nguyen, Phuong, Bowen, Alisa, Edwards, Robert, Stroud, Robert, Zhou, Yi, Van Lookeren Campagne, Menno, and Li, Fei
- Subjects
Mucopolysaccharidosis III ,Humans ,Lysosomes ,Acetyltransferases ,Cryoelectron Microscopy ,Catalytic Domain ,Mutation ,Heparitin Sulfate ,Acetyl Coenzyme A ,Models ,Molecular ,Glucosamine ,Acetylation ,Intracellular Membranes - Abstract
Heparan sulfate (HS) is degraded in lysosome by a series of glycosidases. Before the glycosidases can act, the terminal glucosamine of HS must be acetylated by the integral lysosomal membrane enzyme heparan-α-glucosaminide N-acetyltransferase (HGSNAT). Mutations of HGSNAT cause HS accumulation and consequently mucopolysaccharidosis IIIC, a devastating lysosomal storage disease characterized by progressive neurological deterioration and early death where no treatment is available. HGSNAT catalyzes a unique transmembrane acetylation reaction where the acetyl group of cytosolic acetyl-CoA is transported across the lysosomal membrane and attached to HS in one reaction. However, the reaction mechanism remains elusive. Here we report six cryo-EM structures of HGSNAT along the reaction pathway. These structures reveal a dimer arrangement and a unique structural fold, which enables the elucidation of the reaction mechanism. We find that a central pore within each monomer traverses the membrane and controls access of cytosolic acetyl-CoA to the active site at its luminal mouth where glucosamine binds. A histidine-aspartic acid catalytic dyad catalyzes the transfer reaction via a ternary complex mechanism. Furthermore, the structures allow the mapping of disease-causing variants and reveal their potential impact on the function, thus creating a framework to guide structure-based drug discovery efforts.
- Published
- 2024
9. Structural basis for expanded substrate specificities of human long chain acyl-CoA dehydrogenase and related acyl-CoA dehydrogenases
- Author
-
Narayanan, Beena, Xia, Chuanwu, McAndrew, Ryan, Shen, Anna L, and Kim, Jung-Ja P
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Substrate Specificity ,Humans ,Acyl-CoA Dehydrogenase ,Long-Chain ,Models ,Molecular ,Crystallography ,X-Ray ,Catalytic Domain ,Acyl-CoA Dehydrogenases ,Protein Conformation ,Amino Acid Sequence - Abstract
Crystal structures of human long-chain acyl-CoA dehydrogenase (LCAD) and the catalytically inactive Glu291Gln mutant, have been determined. These structures suggest that LCAD harbors functions beyond its historically defined role in mitochondrial β-oxidation of long and medium-chain fatty acids. LCAD is a homotetramer containing one FAD per 43 kDa subunit with Glu291 as the catalytic base. The substrate binding cavity of LCAD reveals key differences which makes it specific for longer and branched chain substrates. The presence of Pro132 near the start of the E helix leads to helix unwinding that, together with adjacent smaller residues, permits binding of bulky substrates such as 3α, 7α, l2α-trihydroxy-5β-cholestan-26-oyl-CoA. This structural element is also utilized by ACAD11, a eucaryotic ACAD of unknown function, as well as bacterial ACADs known to metabolize sterol substrates. Sequence comparison suggests that ACAD10, another ACAD of unknown function, may also share this substrate specificity. These results suggest that LCAD, ACAD10, ACAD11 constitute a distinct class of eucaryotic acyl CoA dehydrogenases.
- Published
- 2024
10. Structural mechanism of bridge RNA-guided recombination.
- Author
-
Hiraizumi, Masahiro, Perry, Nicholas, Durrant, Matthew, Soma, Teppei, Nagahata, Naoto, Okazaki, Sae, Athukoralage, Januka, Isayama, Yukari, Pai, James, Pawluk, April, Konermann, Silvana, Yamashita, Keitaro, Hsu, Patrick, and Nishimasu, Hiroshi
- Subjects
Recombinases ,DNA ,DNA Transposable Elements ,RNA ,Untranslated ,Cryoelectron Microscopy ,Recombination ,Genetic ,Catalytic Domain ,Nucleic Acid Conformation ,Substrate Specificity ,Models ,Molecular ,Protein Multimerization - Abstract
Insertion sequence (IS) elements are the simplest autonomous transposable elements found in prokaryotic genomes1. We recently discovered that IS110 family elements encode a recombinase and a non-coding bridge RNA (bRNA) that confers modular specificity for target DNA and donor DNA through two programmable loops2. Here we report the cryo-electron microscopy structures of the IS110 recombinase in complex with its bRNA, target DNA and donor DNA in three different stages of the recombination reaction cycle. The IS110 synaptic complex comprises two recombinase dimers, one of which houses the target-binding loop of the bRNA and binds to target DNA, whereas the other coordinates the bRNA donor-binding loop and donor DNA. We uncovered the formation of a composite RuvC-Tnp active site that spans the two dimers, positioning the catalytic serine residues adjacent to the recombination sites in both target and donor DNA. A comparison of the three structures revealed that (1) the top strands of target and donor DNA are cleaved at the composite active sites to form covalent 5-phosphoserine intermediates, (2) the cleaved DNA strands are exchanged and religated to create a Holliday junction intermediate, and (3) this intermediate is subsequently resolved by cleavage of the bottom strands. Overall, this study reveals the mechanism by which a bispecific RNA confers target and donor DNA specificity to IS110 recombinases for programmable DNA recombination.
- Published
- 2024
11. Carotenoid cleavage enzymes evolved convergently to generate the visual chromophore.
- Author
-
Solano, Yasmeen, Dang, Kelly, Abueg, Jude, Kiser, Philip, and Everett, Michael
- Subjects
Carotenoids ,Animals ,Catalytic Domain ,Retinaldehyde ,cis-trans-Isomerases ,Dioxygenases ,Humans ,Models ,Molecular ,Evolution ,Molecular - Abstract
The retinal light response in animals originates from the photoisomerization of an opsin-coupled 11-cis-retinaldehyde chromophore. This visual chromophore is enzymatically produced through the action of carotenoid cleavage dioxygenases. Vertebrates require two carotenoid cleavage dioxygenases, β-carotene oxygenase 1 and retinal pigment epithelium 65 (RPE65), to form 11-cis-retinaldehyde from carotenoid substrates, whereas invertebrates such as insects use a single enzyme known as Neither Inactivation Nor Afterpotential B (NinaB). RPE65 and NinaB couple trans-cis isomerization with hydrolysis and oxygenation, respectively, but the mechanistic relationship of their isomerase activities remains unknown. Here we report the structure of NinaB, revealing details of its active site architecture and mode of membrane binding. Structure-guided mutagenesis studies identify a residue cluster deep within the NinaB substrate-binding cleft that controls its isomerization activity. Our data demonstrate that isomerization activity is mediated by distinct active site regions in NinaB and RPE65-an evolutionary convergence that deepens our understanding of visual system diversity.
- Published
- 2024
12. Antibody discovery identifies regulatory mechanisms of protein arginine deiminase 4.
- Author
-
Zhou, Xin, Kong, Sophie, Maker, Allison, Remesh, Soumya, Leung, Kevin, Verba, Kliment, and Wells, James
- Subjects
Protein-Arginine Deiminase Type 4 ,Humans ,Catalytic Domain ,Cryoelectron Microscopy ,Models ,Molecular ,Antibodies ,Arthritis ,Rheumatoid ,Hydrolases ,Protein-Arginine Deiminases - Abstract
Unlocking the potential of protein arginine deiminase 4 (PAD4) as a drug target for rheumatoid arthritis requires a deeper understanding of its regulation. In this study, we use unbiased antibody selections to identify functional antibodies capable of either activating or inhibiting PAD4 activity. Through cryogenic-electron microscopy, we characterized the structures of these antibodies in complex with PAD4 and revealed insights into their mechanisms of action. Rather than steric occlusion of the substrate-binding catalytic pocket, the antibodies modulate PAD4 activity through interactions with allosteric binding sites adjacent to the catalytic pocket. These binding events lead to either alteration of the active site conformation or the enzyme oligomeric state, resulting in modulation of PAD4 activity. Our study uses antibody engineering to reveal new mechanisms for enzyme regulation and highlights the potential of using PAD4 agonist and antagonist antibodies for studying PAD4-dependency in disease models and future therapeutic development.
- Published
- 2024
13. RNA targeting and cleavage by the type III-Dv CRISPR effector complex.
- Author
-
Schwartz, Evan, Bravo, Jack, Ahsan, Mohd, Macias, Luis, McCafferty, Caitlyn, Dangerfield, Tyler, Walker, Jada, Brodbelt, Jennifer, Fineran, Peter, Fagerlund, Robert, Taylor, David, and Palermo, Giulia
- Subjects
RNA ,RNA ,Catalytic ,CRISPR-Cas Systems ,DNA ,Catalytic Domain ,CRISPR-Associated Proteins ,RNA Cleavage - Abstract
CRISPR-Cas are adaptive immune systems in bacteria and archaea that utilize CRISPR RNA-guided surveillance complexes to target complementary RNA or DNA for destruction1-5. Target RNA cleavage at regular intervals is characteristic of type III effector complexes6-8. Here, we determine the structures of the Synechocystis type III-Dv complex, an apparent evolutionary intermediate from multi-protein to single-protein type III effectors9,10, in pre- and post-cleavage states. The structures show how multi-subunit fusion proteins in the effector are tethered together in an unusual arrangement to assemble into an active and programmable RNA endonuclease and how the effector utilizes a distinct mechanism for target RNA seeding from other type III effectors. Using structural, biochemical, and quantum/classical molecular dynamics simulation, we study the structure and dynamics of the three catalytic sites, where a 2-OH of the ribose on the target RNA acts as a nucleophile for in line self-cleavage of the upstream scissile phosphate. Strikingly, the arrangement at the catalytic residues of most type III complexes resembles the active site of ribozymes, including the hammerhead, pistol, and Varkud satellite ribozymes. Our work provides detailed molecular insight into the mechanisms of RNA targeting and cleavage by an important intermediate in the evolution of type III effector complexes.
- Published
- 2024
14. Network of epistatic interactions in an enzyme active site revealed by large-scale deep mutational scanning
- Author
-
Judge, Allison, Sankaran, Banumathi, Hu, Liya, Palaniappan, Murugesan, Birgy, André, Prasad, BV Venkataram, and Palzkill, Timothy
- Subjects
Biochemistry and Cell Biology ,Bioinformatics and Computational Biology ,Biological Sciences ,Genetics ,Emerging Infectious Diseases ,Antimicrobial Resistance ,Infectious Diseases ,Generic health relevance ,Escherichia coli ,Catalytic Domain ,Mutation ,Amino Acid Substitution ,beta-Lactamases ,cooperativity ,enzyme evolution ,enzyme mechanism ,epistasis ,fitness - Abstract
Cooperative interactions between amino acids are critical for protein function. A genetic reflection of cooperativity is epistasis, which is when a change in the amino acid at one position changes the sequence requirements at another position. To assess epistasis within an enzyme active site, we utilized CTX-M β-lactamase as a model system. CTX-M hydrolyzes β-lactam antibiotics to provide antibiotic resistance, allowing a simple functional selection for rapid sorting of modified enzymes. We created all pairwise mutations across 17 active site positions in the β-lactamase enzyme and quantitated the function of variants against two β-lactam antibiotics using next-generation sequencing. Context-dependent sequence requirements were determined by comparing the antibiotic resistance function of double mutations across the CTX-M active site to their predicted function based on the constituent single mutations, revealing both positive epistasis (synergistic interactions) and negative epistasis (antagonistic interactions) between amino acid substitutions. The resulting trends demonstrate that positive epistasis is present throughout the active site, that epistasis between residues is mediated through substrate interactions, and that residues more tolerant to substitutions serve as generic compensators which are responsible for many cases of positive epistasis. Additionally, we show that a key catalytic residue (Glu166) is amenable to compensatory mutations, and we characterize one such double mutant (E166Y/N170G) that acts by an altered catalytic mechanism. These findings shed light on the unique biochemical factors that drive epistasis within an enzyme active site and will inform enzyme engineering efforts by bridging the gap between amino acid sequence and catalytic function.
- Published
- 2024
15. A bifunctional endolytic alginate lyase with two different lyase catalytic domains from Vibrio sp. H204.
- Author
-
Peng, Chune, Wang, Qingbin, Xu, Wei, Wang, Xinkun, Zheng, Qianqian, Liang, Xiaohui, Dong, Xiaodan, Li, Fuchuan, and Peng, Lizeng
- Subjects
CATALYTIC domains ,POLYSACCHARIDES ,MARINE bacteria ,OLIGOSACCHARIDES ,ALGINIC acid ,ALGINATES - Abstract
Alginate lyases can fully degrade alginate into various size-defined unsaturated oligosaccharide products by β -elimination. Here, we identified the bifunctional endolytic alginate lyase Aly35 from the marine bacterium Vibrio sp. Strain H204. The enzyme Aly35 is classified into the polysaccharide lyase 7 superfamily and contains two alginate lyase catalytic domains. The relationship and function of the two lyase domains are not well known. Thus, the full-length recombinant enzyme and its truncated proteins Aly35-CD1 (catalytic domain 1), Aly35-CD2 (catalytic domain 2 domain) were constructed. The three enzymes showed similar biochemical characteristics and exhibited temperature and pH stability. Further research showed that Aly35 and Aly35-CD2 can efficiently degrade alginate, polymannuronate (PM) and polyguluronate (PG) into a series of unsaturated oligosaccharides, while Aly35-CD1 exhibits greater PM-degrading activity than that of Aly35-CD2 but can not degraded PG efficiently. The results suggest that the domain (Trp
295 -His582 ) is critical for PG-degrading activity, the domain has (Leu53 -Lys286 ) higher PM-degrading activity, both catalytic domains together confer increased alginate (including M-blocks and G blocks)-degrading activity. The enzyme Aly35 and its truncations Aly35-CD1 and Aly35-CD2 will be useful tools for structural analyses and for preparing bioactive oligosaccharides, especially Aly35-CD1 can be used to prepare G unit–rich oligosaccharides from alginate. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
16. Impact of Disease-Associated Mutations on the Deaminase Activity of ADAR1
- Author
-
Karki, Agya, Campbell, Kristen B, Mozumder, Sukanya, Fisher, Andrew J, and Beal, Peter A
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Biomedical and Clinical Sciences ,Genetics ,Rare Diseases ,Pediatric ,Brain Disorders ,2.1 Biological and endogenous factors ,Child ,Humans ,Adenosine Deaminase ,Catalytic Domain ,Mutation ,RNA ,Double-Stranded ,Autoimmune Diseases of the Nervous System ,Nervous System Malformations ,Medicinal and Biomolecular Chemistry ,Biochemistry and Cell Biology ,Medical Biochemistry and Metabolomics ,Biochemistry & Molecular Biology ,Biochemistry and cell biology ,Medical biochemistry and metabolomics ,Medicinal and biomolecular chemistry - Abstract
The innate immune system relies on molecular sensors to detect distinctive molecular patterns, including viral double-stranded RNA (dsRNA), which triggers responses resulting in apoptosis and immune infiltration. Adenosine Deaminases Acting on RNA (ADARs) catalyze the deamination of adenosine (A) to inosine (I), serving as a mechanism to distinguish self from non-self RNA and prevent aberrant immune activation. Loss-of-function mutations in the ADAR1 gene are one cause of Aicardi Goutières Syndrome (AGS), a severe autoimmune disorder in children. Although seven out of the eight AGS-associated mutations in ADAR1 occur within the catalytic domain of the ADAR1 protein, their specific effects on the catalysis of adenosine deamination remain poorly understood. In this study, we carried out a biochemical investigation of four AGS-causing mutations (G1007R, R892H, K999N, and Y1112F) in ADAR1 p110 and truncated variants. These studies included adenosine deamination rate measurements with two different RNA substrates derived from human transcripts known to be edited by ADAR1 p110 (glioma-associated oncogene homologue 1 (hGli1), 5-hydroxytryptamine receptor 2C (5-HT2cR)). Our results indicate that AGS-associated mutations at two amino acid positions directly involved in stabilizing the base-flipped conformation of the ADAR-RNA complex (G1007R and R892H) had the most detrimental impact on catalysis. The K999N mutation, positioned near the RNA binding interface, altered catalysis contextually. Finally, the Y1112F mutation had small effects in each of the assays described here. These findings shed light on the differential effects of disease-associated mutations on adenosine deamination by ADAR1, thereby advancing our structural and functional understanding of ADAR1-mediated RNA editing.
- Published
- 2024
17. Template and target-site recognition by human LINE-1 in retrotransposition.
- Author
-
Thawani, Akanksha, Ariza, Alfredo, Nogales De La Morena, Evangelina, and Collins, Kathleen
- Subjects
Humans ,Cryoelectron Microscopy ,DNA ,Complementary ,Long Interspersed Nucleotide Elements ,Retroelements ,RNA ,Catalytic Domain ,Endonucleases ,Genetic Therapy ,Reverse Transcription ,RNA-Directed DNA Polymerase ,DNA ,Single-Stranded ,DNA Breaks - Abstract
The long interspersed element-1 (LINE-1, hereafter L1) retrotransposon has generated nearly one-third of the human genome and serves as an active source of genetic diversity and human disease1. L1 spreads through a mechanism termed target-primed reverse transcription, in which the encoded enzyme (ORF2p) nicks the target DNA to prime reverse transcription of its own or non-self RNAs2. Here we purified full-length L1 ORF2p and biochemically reconstituted robust target-primed reverse transcription with template RNA and target-site DNA. We report cryo-electron microscopy structures of the complete human L1 ORF2p bound to structured template RNAs and initiating cDNA synthesis. The template polyadenosine tract is recognized in a sequence-specific manner by five distinct domains. Among them, an RNA-binding domain bends the template backbone to allow engagement of an RNA hairpin stem with the L1 ORF2p C-terminal segment. Moreover, structure and biochemical reconstitutions demonstrate an unexpected target-site requirement: L1 ORF2p relies on upstream single-stranded DNA to position the adjacent duplex in the endonuclease active site for nicking of the longer DNA strand, with a single nick generating a staggered DNA break. Our research provides insights into the mechanism of ongoing transposition in the human genome and informs the engineering of retrotransposon proteins for gene therapy.
- Published
- 2024
18. Disordered-to-ordered transitions in assembly factors allow the complex II catalytic subunit to switch binding partners.
- Author
-
Sharma, Pankaj, Maklashina, Elena, Voehler, Markus, Balintova, Sona, Dvorakova, Sarka, Kraus, Michal, Vanova, Katerina, Nahacka, Zuzana, Zobalova, Renata, Boukalova, Stepana, Cunatova, Kristyna, Mracek, Tomas, Ghayee, Hans, Pacak, Karel, Rohlena, Jakub, Neuzil, Jiri, Cecchini, Gary, and Iverson, T
- Subjects
Catalytic Domain ,Protein Structure ,Secondary - Abstract
Complex II (CII) activity controls phenomena that require crosstalk between metabolism and signaling, including neurodegeneration, cancer metabolism, immune activation, and ischemia-reperfusion injury. CII activity can be regulated at the level of assembly, a process that leverages metastable assembly intermediates. The nature of these intermediates and how CII subunits transfer between metastable complexes remains unclear. In this work, we identify metastable species containing the SDHA subunit and its assembly factors, and we assign a preferred temporal sequence of appearance of these species during CII assembly. Structures of two species show that the assembly factors undergo disordered-to-ordered transitions without the appearance of significant secondary structure. The findings identify that intrinsically disordered regions are critical in regulating CII assembly, an observation that has implications for the control of assembly in other biomolecular complexes.
- Published
- 2024
19. Decrypting Allostery in Membrane-Bound K-Ras4B Using Complementary In Silico Approaches Based on Unbiased Molecular Dynamics Simulations.
- Author
-
Castelli, Matteo, Marchetti, Filippo, Osuna, Sílvia, F Oliveira, A, Mulholland, Adrian, Serapian, Stefano, and Colombo, Giorgio
- Subjects
Molecular Dynamics Simulation ,Proteins ,Catalytic Domain ,Guanosine Triphosphate ,Allosteric Regulation - Abstract
Protein functions are dynamically regulated by allostery, which enables conformational communication even between faraway residues, and expresses itself in many forms, akin to different languages: allosteric control pathways predominating in an unperturbed protein are often unintuitively reshaped whenever biochemical perturbations arise (e.g., mutations). To accurately model allostery, unbiased molecular dynamics (MD) simulations require integration with a reliable method able to, e.g., detect incipient allosteric changes or likely perturbation pathways; this is because allostery can operate at longer time scales than those accessible by plain MD. Such methods are typically applied singularly, but we here argue their joint application─as a multilingual approach─could work significantly better. We successfully prove this through unbiased MD simulations (∼100 μs) of the widely studied, allosterically active oncotarget K-Ras4B, solvated and embedded in a phospholipid membrane, from which we decrypt allostery using four showcase languages: Distance Fluctuation analysis and the Shortest Path Map capture allosteric hotspots at equilibrium; Anisotropic Thermal Diffusion and Dynamical Non-Equilibrium MD simulations assess perturbations upon, respectively, either superheating or hydrolyzing the GTP that oncogenically activates K-Ras4B. Chosen languages work synergistically, providing an articulate, mutually coherent, experimentally consistent picture of K-Ras4B allostery, whereby distinct traits emerge at equilibrium and upon GTP cleavage. At equilibrium, combined evidence confirms prominent allosteric communication from the membrane-embedded hypervariable region, through a hub comprising helix α5 and sheet β5, and up to the active site, encompassing allosteric switches I and II (marginally), and two proposed pockets. Upon GTP cleavage, allosteric perturbations mostly accumulate on the switches and documented interfaces.
- Published
- 2024
20. Structure of a ribonucleotide reductase R2 protein radical
- Author
-
Lebrette, Hugo, Srinivas, Vivek, John, Juliane, Aurelius, Oskar, Kumar, Rohit, Lundin, Daniel, Brewster, Aaron S, Bhowmick, Asmit, Sirohiwal, Abhishek, Kim, In-Sik, Gul, Sheraz, Pham, Cindy, Sutherlin, Kyle D, Simon, Philipp, Butryn, Agata, Aller, Pierre, Orville, Allen M, Fuller, Franklin D, Alonso-Mori, Roberto, Batyuk, Alexander, Sauter, Nicholas K, Yachandra, Vittal K, Yano, Junko, Kaila, Ville RI, Sjöberg, Britt-Marie, Kern, Jan, Roos, Katarina, and Högbom, Martin
- Subjects
Biochemistry and Cell Biology ,Chemical Sciences ,Biological Sciences ,Electron Transport ,Protons ,Ribonucleotide Reductases ,Crystallography ,X-Ray ,Entomoplasmataceae ,Catalytic Domain ,Bacterial Proteins ,General Science & Technology - Abstract
Aerobic ribonucleotide reductases (RNRs) initiate synthesis of DNA building blocks by generating a free radical within the R2 subunit; the radical is subsequently shuttled to the catalytic R1 subunit through proton-coupled electron transfer (PCET). We present a high-resolution room temperature structure of the class Ie R2 protein radical captured by x-ray free electron laser serial femtosecond crystallography. The structure reveals conformational reorganization to shield the radical and connect it to the translocation path, with structural changes propagating to the surface where the protein interacts with the catalytic R1 subunit. Restructuring of the hydrogen bond network, including a notably short O-O interaction of 2.41 angstroms, likely tunes and gates the radical during PCET. These structural results help explain radical handling and mobilization in RNR and have general implications for radical transfer in proteins.
- Published
- 2023
21. Interaction of CYP3A4 with caffeine: First insights into multiple substrate binding.
- Author
-
Sevrioukova, Irina
- Subjects
CYP3A4 ,caffeine ,complex ,crystal structure ,cytochrome P450 ,ligand-binding protein ,spectroscopy ,Humans ,Binding Sites ,Caffeine ,Catalytic Domain ,Cytochrome P-450 CYP3A ,Ligands ,Substrate Specificity ,Protein Binding ,Allosteric Regulation ,Crystallography ,X-Ray ,Crystallization ,Demethylation ,Heme ,Hydrophobic and Hydrophilic Interactions ,Mutation - Abstract
Human cytochrome P450 3A4 (CYP3A4) is a major drug-metabolizing enzyme that shows extreme substrate promiscuity. Moreover, its large and malleable active site can simultaneously accommodate several substrate molecules of the same or different nature, which may lead to cooperative binding and allosteric behavior. Due to difficulty of crystallization of CYP3A4-substrate complexes, it remains unknown how multiple substrates can arrange in the active site. We determined crystal structures of CYP3A4 bound to three and six molecules of caffeine, a psychoactive alkaloid serving as a substrate and modulator of CYP3A4. In the ternary complex, one caffeine binds to the active site suitably for C8-hydroxylation, most preferable for CYP3A4. In the senary complex, three caffeine molecules stack parallel to the heme with the proximal ligand poised for 3-N-demethylation. However, the caffeine stack forms extensive hydrophobic interactions that could preclude product dissociation and multiple turnovers. In both complexes, caffeine is also bound in the substrate channel and on the outer surface known as a peripheral site. At all sites, aromatic stacking with the caffeine ring(s) is likely a dominant interaction, while direct and water-mediated polar contacts provide additional stabilization for the substrate-bound complexes. Protein-ligand interactions via the active site R212, intrachannel T224, and peripheral F219 were experimentally confirmed, and the latter two residues were identified as important for caffeine association. Collectively, the structural, spectral, and mutagenesis data provide valuable insights on the ligand binding mechanism and help better understand how purine-based pharmaceuticals and other aromatic compounds could interact with CYP3A4 and mediate drug-drug interactions.
- Published
- 2023
22. A bifunctional endolytic alginate lyase with two different lyase catalytic domains from Vibrio sp. H204
- Author
-
Chune Peng, Qingbin Wang, Wei Xu, Xinkun Wang, Qianqian Zheng, Xiaohui Liang, Xiaodan Dong, Fuchuan Li, and Lizeng Peng
- Subjects
alginate lyase ,bifunctional ,catalytic domain ,oligosaccharides ,marine bacterium ,Microbiology ,QR1-502 - Abstract
Alginate lyases can fully degrade alginate into various size-defined unsaturated oligosaccharide products by β-elimination. Here, we identified the bifunctional endolytic alginate lyase Aly35 from the marine bacterium Vibrio sp. Strain H204. The enzyme Aly35 is classified into the polysaccharide lyase 7 superfamily and contains two alginate lyase catalytic domains. The relationship and function of the two lyase domains are not well known. Thus, the full-length recombinant enzyme and its truncated proteins Aly35-CD1 (catalytic domain 1), Aly35-CD2 (catalytic domain 2 domain) were constructed. The three enzymes showed similar biochemical characteristics and exhibited temperature and pH stability. Further research showed that Aly35 and Aly35-CD2 can efficiently degrade alginate, polymannuronate (PM) and polyguluronate (PG) into a series of unsaturated oligosaccharides, while Aly35-CD1 exhibits greater PM-degrading activity than that of Aly35-CD2 but can not degraded PG efficiently. The results suggest that the domain (Trp295-His582) is critical for PG-degrading activity, the domain has (Leu53-Lys286) higher PM-degrading activity, both catalytic domains together confer increased alginate (including M-blocks and G blocks)-degrading activity. The enzyme Aly35 and its truncations Aly35-CD1 and Aly35-CD2 will be useful tools for structural analyses and for preparing bioactive oligosaccharides, especially Aly35-CD1 can be used to prepare G unit–rich oligosaccharides from alginate.
- Published
- 2024
- Full Text
- View/download PDF
23. The Role of PIK3R1 in Metabolic Function and Insulin Sensitivity.
- Author
-
Tsay, Ariel and Wang, Jen-Chywan
- Subjects
PIK3R1 ,insulin resistance ,metabolic disorders ,p85α ,insulin signaling ,phosphoinositide 3-kinases (PI3K) ,type 2 diabetes ,Animals ,Humans ,Insulin Resistance ,Genes ,Regulator ,Transcription Factors ,Homeostasis ,Catalytic Domain ,Insulin Receptor Substrate Proteins ,Class Ia Phosphatidylinositol 3-Kinase - Abstract
PIK3R1 (also known as p85α) is a regulatory subunit of phosphoinositide 3-kinases (PI3Ks). PI3K, a heterodimer of a regulatory subunit and a catalytic subunit, phosphorylates phosphatidylinositol into secondary signaling molecules involved in regulating metabolic homeostasis. PI3K converts phosphatidylinositol 4,5-bisphosphate (PIP2) to phosphatidylinositol 3,4,5-triphosphate (PIP3), which recruits protein kinase AKT to the inner leaflet of the cell membrane to be activated and to participate in various metabolic functions. PIK3R1 stabilizes and inhibits p110 catalytic activity and serves as an adaptor to interact with insulin receptor substrate (IRS) proteins and growth factor receptors. Thus, mutations in PIK3R1 or altered expression of PIK3R1 could modulate the activity of PI3K and result in significant metabolic outcomes. Interestingly, recent studies also found PI3K-independent functions of PIK3R1. Overall, in this article, we will provide an updated review of the metabolic functions of PIK3R1 that includes studies of PIK3R1 in various metabolic tissues using animal models, the mechanisms modulating PIK3R1 activity, and studies on the mutations of human PIK3R1 gene.
- Published
- 2023
24. Characterization of GEXP15 as a Potential Regulator of Protein Phosphatase 1 in Plasmodium falciparum.
- Author
-
Mansour, Hala, Cabezas-Cruz, Alejandro, Peucelle, Véronique, Farce, Amaury, Salomé-Desnoulez, Sophie, Metatla, Ines, Guerrera, Ida, Hollin, Thomas, and Khalife, Jamal
- Subjects
CD2BP2 ,GEXP15 ,GYF domain ,Plasmodium ,Protein Phosphatase 1 ,malaria ,ribosome biogenesis ,Humans ,Animals ,Plasmodium falciparum ,Protein Phosphatase 1 ,Animals ,Genetically Modified ,Biological Assay ,Catalytic Domain - Abstract
The Protein Phosphatase type 1 catalytic subunit (PP1c) (PF3D7_1414400) operates in combination with various regulatory proteins to specifically direct and control its phosphatase activity. However, there is little information about this phosphatase and its regulators in the human malaria parasite, Plasmodium falciparum. To address this knowledge gap, we conducted a comprehensive investigation into the structural and functional characteristics of a conserved Plasmodium-specific regulator called Gametocyte EXported Protein 15, GEXP15 (PF3D7_1031600). Through in silico analysis, we identified three significant regions of interest in GEXP15: an N-terminal region housing a PP1-interacting RVxF motif, a conserved domain whose function is unknown, and a GYF-like domain that potentially facilitates specific protein-protein interactions. To further elucidate the role of GEXP15, we conducted in vitro interaction studies that demonstrated a direct interaction between GEXP15 and PP1 via the RVxF-binding motif. This interaction was found to enhance the phosphatase activity of PP1. Additionally, utilizing a transgenic GEXP15-tagged line and live microscopy, we observed high expression of GEXP15 in late asexual stages of the parasite, with localization predominantly in the nucleus. Immunoprecipitation assays followed by mass spectrometry analyses revealed the interaction of GEXP15 with ribosomal- and RNA-binding proteins. Furthermore, through pull-down analyses of recombinant functional domains of His-tagged GEXP15, we confirmed its binding to the ribosomal complex via the GYF domain. Collectively, our study sheds light on the PfGEXP15-PP1-ribosome interaction, which plays a crucial role in protein translation. These findings suggest that PfGEXP15 could serve as a potential target for the development of malaria drugs.
- Published
- 2023
25. Mapping roles of active site residues in the acceptor site of the PA3944 Gcn5-related N-acetyltransferase enzyme.
- Author
-
Variot, Cillian, Capule, Daniel, Arolli, Xhulio, Baumgartner, Jackson, Reidl, Cory, Houseman, Charles, Ballicora, Miguel, Becker, Daniel, and Kuhn, Misty
- Subjects
GNAT ,Gcn5-related N-acetyltransferase ,acetylation ,aspartame ,docking visualization ,enzyme kinetics ,molecular docking ,polymyxin B ,substrate docking ,Acetyltransferases ,Catalytic Domain ,Polymyxin B ,Molecular Docking Simulation ,Substrate Specificity ,Kinetics - Abstract
An increased understanding of how the acceptor site in Gcn5-related N-acetyltransferase (GNAT) enzymes recognizes various substrates provides important clues for GNAT functional annotation and their use as chemical tools. In this study, we explored how the PA3944 enzyme from Pseudomonas aeruginosa recognizes three different acceptor substrates, including aspartame, NANMO, and polymyxin B, and identified acceptor residues that are critical for substrate specificity. To achieve this, we performed a series of molecular docking simulations and tested methods to identify acceptor substrate binding modes that are catalytically relevant. We found that traditional selection of best docking poses by lowest S scores did not reveal acceptor substrate binding modes that were generally close enough to the donor for productive acetylation. Instead, sorting poses based on distance between the acceptor amine nitrogen atom and donor carbonyl carbon atom placed these acceptor substrates near residues that contribute to substrate specificity and catalysis. To assess whether these residues are indeed contributors to substrate specificity, we mutated seven amino acid residues to alanine and determined their kinetic parameters. We identified several residues that improved the apparent affinity and catalytic efficiency of PA3944, especially for NANMO and/or polymyxin B. Additionally, one mutant (R106A) exhibited substrate inhibition toward NANMO, and we propose scenarios for the cause of this inhibition based on additional substrate docking studies with R106A. Ultimately, we propose that this residue is a key gatekeeper between the acceptor and donor sites by restricting and orienting the acceptor substrate within the acceptor site.
- Published
- 2023
26. Energetic vs. entropic stabilization between a Remdesivir analogue and cognate ATP upon binding and insertion into the active site of SARS-CoV-2 RNA dependent RNA polymerase
- Author
-
Long, Chunhong, Romero, Moises Ernesto, Dai, Liqiang, and Yu, Jin
- Subjects
Engineering ,Chemical Sciences ,Physical Sciences ,Infectious Diseases ,Emerging Infectious Diseases ,Coronaviruses ,Good Health and Well Being ,Humans ,SARS-CoV-2 ,Catalytic Domain ,RNA ,Viral ,COVID-19 ,COVID-19 Drug Treatment ,Adenosine Monophosphate ,Antiviral Agents ,Adenosine Triphosphate ,Chemical Physics ,Chemical sciences ,Physical sciences - Abstract
SARS-CoV-2 RNA dependent RNA polymerase (RdRp) serves as a highly promising antiviral drug target such as for a Remdesivir nucleotide analogue (RDV-TP or RTP). In this work, we mainly used alchemical all-atom simulations to characterize relative binding free energetics between the nucleotide analogue RTP and natural cognate substrate ATP upon initial binding and pre-catalytic insertion into the active site of SARS-CoV-2 RdRp. Natural non-cognate substrate dATP and mismatched GTP were also examined for computation control. We first identified significant differences in dynamical responses between nucleotide initial binding and subsequent insertion configurations to the open and closed active sites of the RdRp, respectively, though the RdRp protein conformational changes between the active site's open and closed states are subtle. Our alchemical simulations indicated that upon initial binding (active site open), RTP and ATP show similar binding free energies to the active sites while in the insertion state (active site closed), ATP is more stabilized (∼-2.4 kcal mol-1) than RTP in free energetics. Additional analyses show, however, that RTP is more stabilized in binding energetics than ATP, in both the insertion and initial binding states, with RTP more stabilized due to the electrostatic energy in the insertion state and due to vdW energy in the initial binding state. Hence, it appears that natural cognate ATP still excels at association stability with the RdRp active site due to that ATP maintains sufficient flexibilities e.g., in base pairing with the template, which exemplifies an entropic contribution to the cognate substrate stabilization. These findings highlight the importance of substrate flexibilities in addition to energetic stabilization in antiviral nucleotide analogue design.
- Published
- 2023
27. Inverse effects of APOC2 and ANGPTL4 on the conformational dynamics of lid-anchoring structures in lipoprotein lipase.
- Author
-
Kumari, Anni, Grønnemose, Anne, Kristensen, Kristian, Winther, Anne-Marie, Jørgensen, Thomas, Ploug, Michael, and Young, Stephen
- Subjects
ANGPTL4 ,APOC2 ,GPIHBP1 ,HDX-MS ,intravascular lipolysis ,Lipoprotein Lipase ,Angiopoietin-Like Protein 4 ,Apolipoprotein C-II ,Protein Domains ,Catalytic Domain ,Triglycerides - Abstract
The lipolytic processing of triglyceride-rich lipoproteins (TRLs) by lipoprotein lipase (LPL) is crucial for the delivery of dietary lipids to the heart, skeletal muscle, and adipose tissue. The processing of TRLs by LPL is regulated in a tissue-specific manner by a complex interplay between activators and inhibitors. Angiopoietin-like protein 4 (ANGPTL4) inhibits LPL by reducing its thermal stability and catalyzing the irreversible unfolding of LPLs α/β-hydrolase domain. We previously mapped the ANGPTL4 binding site on LPL and defined the downstream unfolding events resulting in LPL inactivation. The binding of LPL to glycosylphosphatidylinositol-anchored high-density lipoprotein-binding protein 1 protects against LPL unfolding. The binding site on LPL for an activating cofactor, apolipoprotein C2 (APOC2), and the mechanisms by which APOC2 activates LPL have been unclear and controversial. Using hydrogen-deuterium exchange/mass spectrometry, we now show that APOC2s C-terminal α-helix binds to regions of LPL surrounding the catalytic pocket. Remarkably, APOC2s binding site on LPL overlaps with that for ANGPTL4, but their effects on LPL conformation are distinct. In contrast to ANGPTL4, APOC2 increases the thermal stability of LPL and protects it from unfolding. Also, the regions of LPL that anchor the lid are stabilized by APOC2 but destabilized by ANGPTL4, providing a plausible explanation for why APOC2 is an activator of LPL, while ANGPTL4 is an inhibitor. Our studies provide fresh insights into the molecular mechanisms by which APOC2 binds and stabilizes LPL-and properties that we suspect are relevant to the conformational gating of LPLs active site.
- Published
- 2023
28. Expanding Extender Substrate Selection for Unnatural Polyketide Biosynthesis by Acyltransferase Domain Exchange within a Modular Polyketide Synthase.
- Author
-
Englund, Elias, Schmidt, Matthias, Nava, Alberto A, Lechner, Anna, Deng, Kai, Jocic, Renee, Lin, Yingxin, Roberts, Jacob, Benites, Veronica T, Kakumanu, Ramu, Gin, Jennifer W, Chen, Yan, Liu, Yuzhong, Petzold, Christopher J, Baidoo, Edward EK, Northen, Trent R, Adams, Paul D, Katz, Leonard, Yuzawa, Satoshi, and Keasling, Jay D
- Subjects
Polyketide Synthases ,Acyltransferases ,Catalytic Domain ,Substrate Specificity ,Polyketides ,Generic health relevance ,Chemical Sciences ,General Chemistry - Abstract
Modular polyketide synthases (PKSs) are polymerases that employ α-carboxyacyl-CoAs as extender substrates. This enzyme family contains several catalytic modules, where each module is responsible for a single round of polyketide chain extension. Although PKS modules typically use malonyl-CoA or methylmalonyl-CoA for chain elongation, many other malonyl-CoA analogues are used to diversify polyketide structures in nature. Previously, we developed a method to alter an extension substrate of a given module by exchanging an acyltransferase (AT) domain while maintaining protein folding. Here, we report in vitro polyketide biosynthesis by 13 PKSs (the wild-type PKS and 12 AT-exchanged PKSs with unusual ATs) and 14 extender substrates. Our ∼200 in vitro reactions resulted in 13 structurally different polyketides, including several polyketides that have not been reported. In some cases, AT-exchanged PKSs produced target polyketides by >100-fold compared to the wild-type PKS. These data also indicate that most unusual AT domains do not incorporate malonyl-CoA and methylmalonyl-CoA but incorporate various rare extender substrates that are equal to in size or slightly larger than natural substrates. We developed a computational workflow to predict the approximate AT substrate range based on active site volumes to support the selection of ATs. These results greatly enhance our understanding of rare AT domains and demonstrate the benefit of using the proposed PKS engineering strategy to produce novel chemicals in vitro.
- Published
- 2023
29. Metalloallostery and Transition Metal Signaling: Bioinorganic Copper Chemistry Beyond Active Sites.
- Author
-
Pham, Vanha and Chang, Christopher
- Subjects
Copper Fluorescent Sensor ,Cuproplasia ,Cuproptosis ,Metalloallostery ,Transition Metal Signaling ,Copper ,Catalytic Domain ,Chemistry ,Bioinorganic ,Transition Elements ,Metals ,Proteins ,Binding Sites - Abstract
Transition metal chemistry is essential to life, where metal binding to DNA, RNA, and proteins underpins all facets of the central dogma of biology. In this context, metals in proteins are typically studied as static active site cofactors. However, the emergence of transition metal signaling, where mobile metal pools can transiently bind to biological targets beyond active sites, is expanding this conventional view of bioinorganic chemistry. This Minireview focuses on the concept of metalloallostery, using copper as a canonical example of how metals can regulate protein function by binding to remote allosteric sites (e.g., exosites). We summarize advances in and prospects for the field, including imaging dynamic transition metal signaling pools, allosteric inhibition or activation of protein targets by metal binding, and metal-dependent signaling pathways that underlie nutrient vulnerabilities in diseases spanning obesity, fatty liver disease, cancer, and neurodegeneration.
- Published
- 2023
30. 2,5-Pyridinedicarboxylic acid is a bioactive and highly selective inhibitor of D-dopachrome tautomerase
- Author
-
Parkins, Andrew, Das, Pragnya, Prahaladan, Varsha, Rangel, Vanessa M, Xue, Liang, Sankaran, Banumathi, Bhandari, Vineet, and Pantouris, Georgios
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,5.1 Pharmaceuticals ,Humans ,Catalytic Domain ,CD74 ,D-dopachrome tautomerase ,active site volume ,bioactive ,inhibitor ,selectivity ,Chemical Sciences ,Information and Computing Sciences ,Biophysics ,Biological sciences ,Chemical sciences - Abstract
Macrophage migration inhibitory factor (MIF) and D-dopachrome tautomerase (D-DT) are two pleotropic cytokines, which are coexpressed in various cell types to activate the cell surface receptor CD74. Via the MIF/CD74 and D-DT/CD74 axes, the two proteins exhibit either beneficial or deleterious effect on human diseases. In this study, we report the identification of 2,5-pyridinedicarboxylic acid (a.k.a. 1) that effectively blocks the D-DT-induced activation of CD74 and demonstrates an impressive 79-fold selectivity for D-DT over MIF. Crystallographic characterization of D-DT-1 elucidates the binding features of 1 and reveals previously unrecognized differences between the MIF and D-DT active sites that explain the ligand's functional selectivity. The commercial availability, low cost, and high selectivity make 1 the ideal tool for studying the pathophysiological functionality of D-DT in disease models. At the same time, our comprehensive biochemical, computational, and crystallographic analyses serve as a guide for generating highly potent and selective D-DT inhibitors.
- Published
- 2023
31. Structure of the lysosomal mTORC1–TFEB–Rag–Ragulator megacomplex
- Author
-
Cui, Zhicheng, Napolitano, Gennaro, de Araujo, Mariana EG, Esposito, Alessandra, Monfregola, Jlenia, Huber, Lukas A, Ballabio, Andrea, and Hurley, James H
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Genetics ,1.1 Normal biological development and functioning ,Generic health relevance ,Amino Acids ,Catalytic Domain ,Guanosine Diphosphate ,Lysosomes ,Mechanistic Target of Rapamycin Complex 1 ,Monomeric GTP-Binding Proteins ,Phosphorylation ,Protein Multimerization ,Regulatory-Associated Protein of mTOR ,Signal Transduction ,General Science & Technology - Abstract
The transcription factor TFEB is a master regulator of lysosomal biogenesis and autophagy1. The phosphorylation of TFEB by the mechanistic target of rapamycin complex 1 (mTORC1)2-5 is unique in its mTORC1 substrate recruitment mechanism, which is strictly dependent on the amino acid-mediated activation of the RagC GTPase activating protein FLCN6,7. TFEB lacks the TOR signalling motif responsible for the recruitment of other mTORC1 substrates. We used cryogenic-electron microscopy to determine the structure of TFEB as presented to mTORC1 for phosphorylation, which we refer to as the 'megacomplex'. Two full Rag-Ragulator complexes present each molecule of TFEB to the mTOR active site. One Rag-Ragulator complex is bound to Raptor in the canonical mode seen previously in the absence of TFEB. A second Rag-Ragulator complex (non-canonical) docks onto the first through a RagC GDP-dependent contact with the second Ragulator complex. The non-canonical Rag dimer binds the first helix of TFEB with a RagCGDP-dependent aspartate clamp in the cleft between the Rag G domains. In cellulo mutation of the clamp drives TFEB constitutively into the nucleus while having no effect on mTORC1 localization. The remainder of the 108-amino acid TFEB docking domain winds around Raptor and then back to RagA. The double use of RagC GDP contacts in both Rag dimers explains the strong dependence of TFEB phosphorylation on FLCN and the RagC GDP state.
- Published
- 2023
32. Catalytic site mutations confer multiple states of G protein activation.
- Author
-
Hewitt, Natalie, Ma, Ning, Arang, Nadia, Martin, Sarah, Prakash, Ajit, DiBerto, Jeffrey, Knight, Kevin, Ghosh, Soumadwip, Olsen, Reid, Roth, Bryan, Gutkind, J, Vaidehi, Nagarajan, Campbell, Sharon, and Dohlman, Henrik
- Subjects
Catalytic Domain ,Glutamine ,Heterotrimeric GTP-Binding Proteins ,Mutation ,Guanosine Triphosphate - Abstract
Heterotrimeric guanine nucleotide-binding proteins (G proteins) that function as molecular switches for cellular growth and metabolism are activated by GTP and inactivated by GTP hydrolysis. In uveal melanoma, a conserved glutamine residue critical for GTP hydrolysis in the G protein α subunit is often mutated in Gαq or Gα11 to either leucine or proline. In contrast, other glutamine mutations or mutations in other Gα subtypes are rare. To uncover the mechanism of the genetic selection and the functional role of this glutamine residue, we analyzed all possible substitutions of this residue in multiple Gα isoforms. Through cell-based measurements of activity, we showed that some mutants were further activated and inactivated by G protein-coupled receptors. Through biochemical, molecular dynamics, and nuclear magnetic resonance-based structural studies, we showed that the Gα mutants were functionally distinct and conformationally diverse, despite their shared inability to hydrolyze GTP. Thus, the catalytic glutamine residue contributes to functions beyond GTP hydrolysis, and these functions include subtype-specific, allosteric modulation of receptor-mediated subunit dissociation. We conclude that G proteins do not function as simple on-off switches. Rather, signaling emerges from an ensemble of active states, a subset of which are favored in disease and may be uniquely responsive to receptor-directed ligands.
- Published
- 2023
33. Geometry of Charge Density as a Reporter on the Role of the Protein Scaffold in Enzymatic Catalysis: Electrostatic Preorganization and Beyond
- Author
-
Eberhart, Mark E, Wilson, Timothy R, Johnston, Nathaniel W, and Alexandrova, Anastassia N
- Subjects
Chemical Sciences ,Physical Chemistry ,Theoretical and Computational Chemistry ,Bioengineering ,Static Electricity ,Catalysis ,Catalytic Domain ,Motion ,Biochemistry and Cell Biology ,Computer Software ,Chemical Physics ,Physical chemistry ,Theoretical and computational chemistry - Abstract
Enzymes host active sites inside protein macromolecules, which have diverse, often incredibly complex, and atom-expensive structures. It is an outstanding question what the role of these expensive scaffolds might be in enzymatic catalysis. Answering this question is essential to both enzymology and the design of artificial enzymes with proficiencies that will match those of the best natural enzymes. Protein rigidifying the active site, contrasted with the dynamics and vibrational motion promoting the reaction, as well as long-range electrostatics (also known as electrostatic preorganization) were all proposed as central contributions of the scaffold to the catalysis. Here, we show that all these effects inevitably produce changes in the quantum mechanical electron density in the active site, which in turn defines the reactivity. The phenomena are therefore fundamentally inseparable. The geometry of the electron density-a scalar field characterized by a number of mathematical features such as critical points-is a rigorous and convenient descriptor of enzymatic catalysis and a reporter on the role of the protein. We show how this geometry can be analyzed, linked to the reaction barriers, and report in particular on intramolecular electric fields in enzymes. We illustrate these tools on the studies of electrostatic preorganization in several representative enzyme classes, both natural and artificial. We highlight the forward-looking aspects of the approach.
- Published
- 2023
34. De novo design of luciferases using deep learning.
- Author
-
Yeh, Andy Hsien-Wei, Norn, Christoffer, Kipnis, Yakov, Tischer, Doug, Pellock, Samuel J, Evans, Declan, Ma, Pengchen, Lee, Gyu Rie, Zhang, Jason Z, Anishchenko, Ivan, Coventry, Brian, Cao, Longxing, Dauparas, Justas, Halabiya, Samer, DeWitt, Michelle, Carter, Lauren, Houk, KN, and Baker, David
- Subjects
Luciferases ,Enzyme Stability ,Catalytic Domain ,Substrate Specificity ,Oxidation-Reduction ,Luminescence ,Hot Temperature ,Biocatalysis ,Deep Learning ,Luciferins ,Biotechnology ,Generic health relevance ,General Science & Technology - Abstract
De novo enzyme design has sought to introduce active sites and substrate-binding pockets that are predicted to catalyse a reaction of interest into geometrically compatible native scaffolds1,2, but has been limited by a lack of suitable protein structures and the complexity of native protein sequence-structure relationships. Here we describe a deep-learning-based 'family-wide hallucination' approach that generates large numbers of idealized protein structures containing diverse pocket shapes and designed sequences that encode them. We use these scaffolds to design artificial luciferases that selectively catalyse the oxidative chemiluminescence of the synthetic luciferin substrates diphenylterazine3 and 2-deoxycoelenterazine. The designed active sites position an arginine guanidinium group adjacent to an anion that develops during the reaction in a binding pocket with high shape complementarity. For both luciferin substrates, we obtain designed luciferases with high selectivity; the most active of these is a small (13.9 kDa) and thermostable (with a melting temperature higher than 95 °C) enzyme that has a catalytic efficiency on diphenylterazine (kcat/Km = 106 M-1 s-1) comparable to that of native luciferases, but a much higher substrate specificity. The creation of highly active and specific biocatalysts from scratch with broad applications in biomedicine is a key milestone for computational enzyme design, and our approach should enable generation of a wide range of luciferases and other enzymes.
- Published
- 2023
35. Bond Bundle Analysis of Ketosteroid Isomerase
- Author
-
Wilson, Timothy R, Morgenstern, Amanda, Alexandrova, Anastassia N, and Eberhart, ME
- Subjects
Chemical Sciences ,Physical Chemistry ,Steroid Isomerases ,Hydrogen Bonding ,Ketosteroids ,Catalytic Domain ,Catalysis ,Isomerases ,Physical Sciences ,Engineering ,Chemical sciences ,Physical sciences - Abstract
Bond bundle analysis is used to investigate enzymatic catalysis in the ketosteroid isomerase (KSI) active site. We identify the unique bonding regions in five KSI systems, including those exposed to applied oriented electric fields and those with amino acid mutations, and calculate the precise redistribution of electron density and other regional properties that accompanies either enhancement or inhibition of KSI catalytic activity. We find that catalytic enhancement results from promoting both inter- and intra-molecular electron density redistribution, between bond bundles and bond wedges within the KSI-docked substrate molecule, in the forward direction of the catalyzed reaction. Though the redistribution applies to both types of perturbed systems and is thus suggestive of a general catalytic role, we observe that bond properties (e.g., volume vs energy vs electron count) can respond independently and disproportionately depending on the type of perturbation. We conclude that the resulting catalytic enhancement/inhibition proceeds via different mechanisms, where some bond properties are utilized more by one type of perturbation than the other. Additionally, we find that the correlations between bond wedge properties and catalyzed reaction barrier energies are additive to predict those of bond bundles and atomic basins, providing a rigorous grounding for connecting changes in local charge density to resulting shifts in reaction barrier energy.
- Published
- 2022
36. Early Nitrogenase Ancestors Encompassed Novel Active Site Diversity
- Author
-
Schwartz, Sarah L, Garcia, Amanda K, Kaçar, Betül, and Fournier, Gregory P
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Prevention ,Nitrogenase ,Catalytic Domain ,Proteins ,Substrate Specificity ,Phylogeny ,ancestral sequence reconstruction ,nitrogenase ,Nif ,early life ,Evolutionary Biology ,Genetics ,Biochemistry and cell biology ,Evolutionary biology - Abstract
Ancestral sequence reconstruction (ASR) infers predicted ancestral states for sites within sequences and can constrain the functions and properties of ancestors of extant protein families. Here, we compare the likely sequences of inferred nitrogenase ancestors to extant nitrogenase sequence diversity. We show that the most-likely combinations of ancestral states for key substrate channel residues are not represented in extant sequence space, and rarely found within a more broadly defined physiochemical space-supporting that the earliest ancestors of extant nitrogenases likely had alternative substrate channel composition. These differences may indicate differing environmental selection pressures acting on nitrogenase substrate specificity in ancient environments. These results highlight ASR's potential as an in silico tool for developing hypotheses about ancestral enzyme functions, as well as improving hypothesis testing through more targeted in vitro and in vivo experiments.
- Published
- 2022
37. An active site loop toggles between conformations to control antibiotic hydrolysis and inhibition potency for CTX-M β-lactamase drug-resistance enzymes
- Author
-
Lu, Shuo, Hu, Liya, Lin, Hanfeng, Judge, Allison, Rivera, Paola, Palaniappan, Murugesan, Sankaran, Banumathi, Wang, Jin, Prasad, BV Venkataram, and Palzkill, Timothy
- Subjects
Biochemistry and Cell Biology ,Medical Microbiology ,Biomedical and Clinical Sciences ,Biological Sciences ,Biodefense ,Infectious Diseases ,Antimicrobial Resistance ,Emerging Infectious Diseases ,5.1 Pharmaceuticals ,Anti-Bacterial Agents ,Catalytic Domain ,Hydrolysis ,Escherichia coli ,beta-Lactamases - Abstract
β-lactamases inactivate β-lactam antibiotics leading to drug resistance. Consequently, inhibitors of β-lactamases can combat this resistance, and the β-lactamase inhibitory protein (BLIP) is a naturally occurring inhibitor. The widespread CTX-M-14 and CTX-M-15 β-lactamases have an 83% sequence identity. In this study, we show that BLIP weakly inhibits CTX-M-14 but potently inhibits CTX-M-15. The structure of the BLIP/CTX-M-15 complex reveals that binding is associated with a conformational change of an active site loop of β-lactamase. Surprisingly, the loop structure in the complex is similar to that in a drug-resistant variant (N106S) of CTX-M-14. We hypothesized that the pre-established favorable loop conformation of the N106S mutant would facilitate binding. The N106S substitution results in a ~100- and 10-fold increase in BLIP inhibition potency for CTX-M-14 and CTX-M-15, respectively. Thus, this indicates that an active site loop in β-lactamase toggles between conformations that control antibiotic hydrolysis and inhibitor susceptibility. These findings highlight the role of accessible active site conformations in controlling enzyme activity and inhibitor susceptibility as well as the influence of mutations in selectively stabilizing discrete conformations.
- Published
- 2022
38. Essential Role of Loop Dynamics in Type II NRPS Biomolecular Recognition
- Author
-
Corpuz, Joshua C, Patel, Ashay, Davis, Tony D, Podust, Larissa M, McCammon, J Andrew, and Burkart, Michael D
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Bioengineering ,Generic health relevance ,Amino Acid Sequence ,Peptide Synthases ,Peptides ,Catalytic Domain ,Carrier Proteins ,Chemical Sciences ,Organic Chemistry ,Biological sciences ,Chemical sciences - Abstract
Non-ribosomal peptides play a critical role in the clinic as therapeutic agents. To access more chemically diverse therapeutics, non-ribosomal peptide synthetases (NRPSs) have been targeted for engineering through combinatorial biosynthesis; however, this has been met with limited success in part due to the lack of proper protein-protein interactions between non-cognate proteins. Herein, we report our use of chemical biology to enable X-ray crystallography, molecular dynamics (MD) simulations, and biochemical studies to elucidate binding specificities between peptidyl carrier proteins (PCPs) and adenylation (A) domains. Specifically, we determined X-ray crystal structures of a type II PCP crosslinked to its cognate A domain, PigG and PigI, and of PigG crosslinked to a non-cognate PigI homologue, PltF. The crosslinked PCP-A domain structures possess large protein-protein interfaces that predominantly feature hydrophobic interactions, with specific electrostatic interactions that orient the substrate for active site delivery. MD simulations of the PCP-A domain complexes and unbound PCP structures provide a dynamical evaluation of the transient interactions formed at PCP-A domain interfaces, which confirm the previously hypothesized role of a PCP loop as a crucial recognition element. Finally, we demonstrate that the interfacial interactions at the PCP loop 1 region can be modified to control PCP binding specificity through gain-of-function mutations. This work suggests that loop conformational preferences and dynamism account for improved shape complementary in the PCP-A domain interactions. Ultimately, these studies show how crystallographic, biochemical, and computational methods can be used to rationally re-engineer NRPSs for non-cognate interactions.
- Published
- 2022
39. Genetic variants associated with psychiatric disorders are enriched at epigenetically active sites in lymphoid cells.
- Author
-
Lynall, Mary-Ellen, Soskic, Blagoje, Hayhurst, James, Schwartzentruber, Jeremy, Levey, Daniel, Pathak, Gita, Polimanti, Renato, Gelernter, Joel, Stein, Murray, Trynka, Gosia, Clatworthy, Menna, and Bullmore, Ed
- Subjects
Catalytic Domain ,Genetic Predisposition to Disease ,Genome-Wide Association Study ,Humans ,Lymphocytes ,Mental Disorders ,Polymorphism ,Single Nucleotide ,Schizophrenia - Abstract
Multiple psychiatric disorders have been associated with abnormalities in both the innate and adaptive immune systems. The role of these abnormalities in pathogenesis, and whether they are driven by psychiatric risk variants, remains unclear. We test for enrichment of GWAS variants associated with multiple psychiatric disorders (cross-disorder or trans-diagnostic risk), or 5 specific disorders (cis-diagnostic risk), in regulatory elements in immune cells. We use three independent epigenetic datasets representing multiple organ systems and immune cell subsets. Trans-diagnostic and cis-diagnostic risk variants (for schizophrenia and depression) are enriched at epigenetically active sites in brain tissues and in lymphoid cells, especially stimulated CD4+ T cells. There is no evidence for enrichment of either trans-risk or cis-risk variants for schizophrenia or depression in myeloid cells. This suggests a possible model where environmental stimuli activate T cells to unmask the effects of psychiatric risk variants, contributing to the pathogenesis of mental health disorders.
- Published
- 2022
40. Ir(III)-Based Agents for Monitoring the Cytochrome P450 3A4 Active Site Occupancy.
- Author
-
Denison, Madeline, Steinke, Sean, Majeed, Aliza, Turro, Claudia, Kocarek, Thomas, Sevrioukova, Irina, and Kodanko, Jeremy
- Subjects
Catalytic Domain ,Cytochrome P-450 CYP3A ,Cytochrome P-450 Enzyme System ,Heme ,Humans ,Iridium - Abstract
Cytochromes P450 (CYPs) are a superfamily of enzymes responsible for biosynthesis and drug metabolism. Monitoring the activity of CYP3A4, the major human drug-metabolizing enzyme, is vital for assessing the metabolism of pharmaceuticals and identifying harmful drug-drug interactions. Existing probes for CYP3A4 are irreversible turn-on substrates that monitor activity at specific time points in end-point assays. To provide a more dynamic approach, we designed, synthesized, and characterized emissive Ir(III) and Ru(II) complexes that allow monitoring of the CYP3A4 active-site occupancy in real time. In the bound state, probe emission is quenched by the active-site heme. Upon displacement from the active site by CYP3A4-specific inhibitors or substrates, these probes show high emission turn-on. Direct probe binding to the CYP3A4 active site was confirmed by X-ray crystallography. The lead Ir(III)-based probe has nanomolar Kd and high selectivity for CYP3A4, efficient cellular uptake, and low toxicity in CYP3A4-overexpressing HepG2 cells.
- Published
- 2022
41. Functional roles of enzyme dynamics in accelerating active site chemistry: Emerging techniques and changing concepts.
- Author
-
Gao, Shuaihua and Klinman, Judith
- Subjects
Biophysics ,Catalysis ,Catalytic Domain ,Protein Conformation ,Proteins - Abstract
With the growing acceptance of the contribution of protein conformational ensembles to enzyme catalysis and regulation, research in the field of protein dynamics has shifted toward an understanding of the atomistic properties of protein dynamical networks and the mechanisms and time scales that control such behavior. A full description of an enzymatic reaction coordinate is expected to extend beyond the active site and include site-specific networks that communicate with the protein/water interface. Advances in experimental tools for the spatial resolution of thermal activation pathways are being complemented by biophysical methods for visualizing dynamics in real time. An emerging multidimensional model integrates the impacts of bound substrate/effector on the distribution of protein substates that are in rapid equilibration near room temperature with reaction-specific protein embedded heat transfer conduits.
- Published
- 2022
42. Theoretical Perspective on Operando Spectroscopy of Fluxional Nanocatalysts
- Author
-
Poths, Patricia and Alexandrova, Anastassia N
- Subjects
Catalysis ,Catalytic Domain ,Spectrum Analysis ,Physical Sciences ,Chemical Sciences - Abstract
Improvements in operando spectroscopy have enabled the catalysis community to investigate the dynamic nature of catalysts under operating conditions with increasing detail. Still, the highly dynamic nature of some catalysts, such as fluxional supported subnano clusters, presents a formidable challenge even for the most state-of-the-art techniques. The reason is that such fluxional catalytic interfaces contain a variety of thermally accessible states. Operando spectroscopies used in catalysis generally fall into two categories: ensemble-based techniques, which provide spectra containing the signals of the entire ensemble of states of the catalyst and are not necessarily dominated by the most active species, and localized techniques, which provide atomistic-level information about the dynamics of active sites in a very small area, which might not include the most active species. Combining many different kinds of techniques can provide detailed insight; however, we propose that effective utilization of specific computational techniques and approaches within the fluxionality paradigm can fill the gap and enable atomistic characterization of the most relevant catalytic sites.
- Published
- 2022
43. Dilute Alloys Based on Au, Ag, or Cu for Efficient Catalysis: From Synthesis to Active Sites
- Author
-
Lee, Jennifer D, Miller, Jeffrey B, Shneidman, Anna V, Sun, Lixin, Weaver, Jason F, Aizenberg, Joanna, Biener, Juergen, Boscoboinik, J Anibal, Foucher, Alexandre C, Frenkel, Anatoly I, van der Hoeven, Jessi ES, Kozinsky, Boris, Marcella, Nicholas, Montemore, Matthew M, Ngan, Hio Tong, O’Connor, Christopher R, Owen, Cameron J, Stacchiola, Dario J, Stach, Eric A, Madix, Robert J, Sautet, Philippe, and Friend, Cynthia M
- Subjects
Affordable and Clean Energy ,Alloys ,Catalysis ,Catalytic Domain ,Metals ,Oxidation-Reduction ,Oxides ,Chemical Sciences ,General Chemistry - Abstract
The development of new catalyst materials for energy-efficient chemical synthesis is critical as over 80% of industrial processes rely on catalysts, with many of the most energy-intensive processes specifically using heterogeneous catalysis. Catalytic performance is a complex interplay of phenomena involving temperature, pressure, gas composition, surface composition, and structure over multiple length and time scales. In response to this complexity, the integrated approach to heterogeneous dilute alloy catalysis reviewed here brings together materials synthesis, mechanistic surface chemistry, reaction kinetics, in situ and operando characterization, and theoretical calculations in a coordinated effort to develop design principles to predict and improve catalytic selectivity. Dilute alloy catalysts─in which isolated atoms or small ensembles of the minority metal on the host metal lead to enhanced reactivity while retaining selectivity─are particularly promising as selective catalysts. Several dilute alloy materials using Au, Ag, and Cu as the majority host element, including more recently introduced support-free nanoporous metals and oxide-supported nanoparticle "raspberry colloid templated (RCT)" materials, are reviewed for selective oxidation and hydrogenation reactions. Progress in understanding how such dilute alloy catalysts can be used to enhance selectivity of key synthetic reactions is reviewed, including quantitative scaling from model studies to catalytic conditions. The dynamic evolution of catalyst structure and composition studied in surface science and catalytic conditions and their relationship to catalytic function are also discussed, followed by advanced characterization and theoretical modeling that have been developed to determine the distribution of minority metal atoms at or near the surface. The integrated approach demonstrates the success of bridging the divide between fundamental knowledge and design of catalytic processes in complex catalytic systems, which can accelerate the development of new and efficient catalytic processes.
- Published
- 2022
44. Overcoming universal restrictions on metal selectivity by protein design
- Author
-
Choi, Tae Su and Tezcan, F Akif
- Subjects
Inorganic Chemistry ,Biochemistry and Cell Biology ,Chemical Sciences ,Biological Sciences ,Amino Acids ,Catalytic Domain ,Ions ,Metalloproteins ,Metals ,Proteins ,General Science & Technology - Abstract
Selective metal coordination is central to the functions of metalloproteins:1,2 each metalloprotein must pair with its cognate metallocofactor to fulfil its biological role3. However, achieving metal selectivity solely through a three-dimensional protein structure is a great challenge, because there is a limited set of metal-coordinating amino acid functionalities and proteins are inherently flexible, which impedes steric selection of metals3,4. Metal-binding affinities of natural proteins are primarily dictated by the electronic properties of metal ions and follow the Irving-Williams series5 (Mn2+ Zn2+) with few exceptions6,7. Accordingly, metalloproteins overwhelmingly bind Cu2+ and Zn2+ in isolation, regardless of the nature of their active sites and their cognate metal ions1,3,8. This led organisms to evolve complex homeostatic machinery and non-equilibrium strategies to achieve correct metal speciation1,3,8-10. Here we report an artificial dimeric protein, (AB)2, that thermodynamically overcomes the Irving-Williams restrictions in vitro and in cells, favouring the binding of lower-Irving-Williams transition metals over Cu2+, the most dominant ion in the Irving-Williams series. Counter to the convention in molecular design of achieving specificity through structural preorganization, (AB)2 was deliberately designed to be flexible. This flexibility enabled (AB)2 to adopt mutually exclusive, metal-dependent conformational states, which led to the discovery of structurally coupled coordination sites that disfavour Cu2+ ions by enforcing an unfavourable coordination geometry. Aside from highlighting flexibility as a valuable element in protein design, our results illustrate design principles for constructing selective metal sequestration agents.
- Published
- 2022
45. Structure and dynamics of SARS-CoV-2 proofreading exoribonuclease ExoN
- Author
-
Moeller, Nicholas H, Shi, Ke, Demir, Özlem, Belica, Christopher, Banerjee, Surajit, Yin, Lulu, Durfee, Cameron, Amaro, Rommie E, and Aihara, Hideki
- Subjects
Biodefense ,Genetics ,Pneumonia ,Prevention ,Pneumonia & Influenza ,Emerging Infectious Diseases ,Vaccine Related ,Infectious Diseases ,Lung ,Infection ,Binding Sites ,COVID-19 ,Catalytic Domain ,Crystallography ,X-Ray ,Exoribonucleases ,Humans ,Lysine ,Molecular Dynamics Simulation ,Mutation ,Missense ,Nucleic Acid Conformation ,Protein Binding ,Protein Domains ,RNA ,Viral ,SARS-CoV-2 ,Viral Nonstructural Proteins ,exoribonuclease ,proofreading ,molecular dynamics simulations ,crystal structure - Abstract
High-fidelity replication of the large RNA genome of coronaviruses (CoVs) is mediated by a 3'-to-5' exoribonuclease (ExoN) in nonstructural protein 14 (nsp14), which excises nucleotides including antiviral drugs misincorporated by the low-fidelity viral RNA-dependent RNA polymerase (RdRp) and has also been implicated in viral RNA recombination and resistance to innate immunity. Here, we determined a 1.6-Å resolution crystal structure of severe acute respiratory syndrome CoV 2 (SARS-CoV-2) ExoN in complex with its essential cofactor, nsp10. The structure shows a highly basic and concave surface flanking the active site, comprising several Lys residues of nsp14 and the N-terminal amino group of nsp10. Modeling suggests that this basic patch binds to the template strand of double-stranded RNA substrates to position the 3' end of the nascent strand in the ExoN active site, which is corroborated by mutational and computational analyses. We also show that the ExoN activity can rescue a stalled RNA primer poisoned with sofosbuvir and allow RdRp to continue its extension in the presence of the chain-terminating drug, biochemically recapitulating proofreading in SARS-CoV-2 replication. Molecular dynamics simulations further show remarkable flexibility of multidomain nsp14 and suggest that nsp10 stabilizes ExoN for substrate RNA binding to support its exonuclease activity. Our high-resolution structure of the SARS-CoV-2 ExoN-nsp10 complex serves as a platform for future development of anticoronaviral drugs or strategies to attenuate the viral virulence.
- Published
- 2022
46. Organometallic Fe2(μ-SH)2(CO)4(CN)2 Cluster Allows the Biosynthesis of the [FeFe]-Hydrogenase with Only the HydF Maturase
- Author
-
Zhang, Yu, Tao, Lizhi, Woods, Toby J, Britt, R David, and Rauchfuss, Thomas B
- Subjects
Inorganic Chemistry ,Chemical Sciences ,Bacterial Proteins ,Catalysis ,Catalytic Domain ,Electron Spin Resonance Spectroscopy ,Hydrogen ,Hydrogenase ,Iron-Sulfur Proteins ,Molecular Conformation ,Organometallic Compounds ,Oxidation-Reduction ,Trans-Activators ,General Chemistry ,Chemical sciences ,Engineering - Abstract
The biosynthesis of the active site of the [FeFe]-hydrogenases (HydA1), the H-cluster, is of interest because these enzymes are highly efficient catalysts for the oxidation and production of H2. The biosynthesis of the [2Fe]H subcluster of the H-cluster proceeds from simple precursors, which are processed by three maturases: HydG, HydE, and HydF. Previous studies established that HydG produces an Fe(CO)2(CN) adduct of cysteine, which is the substrate for HydE. In this work, we show that by using the synthetic cluster [Fe2(μ-SH)2(CN)2(CO)4]2- active HydA1 can be biosynthesized without maturases HydG and HydE.
- Published
- 2022
47. Updating the Paradigm: Redox Partner Binding and Conformational Dynamics in Cytochromes P450.
- Author
-
Poulos, Thomas and Follmer, Alec
- Subjects
Binding Sites ,Camphor 5-Monooxygenase ,Catalytic Domain ,Cytochrome P-450 Enzyme System ,Ferredoxins ,Humans ,Molecular Dynamics Simulation ,Oxidation-Reduction ,Protein Conformation - Abstract
This Account summarizes recent findings centered on the role that redox partner binding, allostery, and conformational dynamics plays in cytochrome P450 proton coupled electron transfer. P450s are one of Natures largest enzyme families and it is not uncommon to find a P450 wherever substrate oxidation is required in the formation of essential molecules critical to the life of the organism or in xenobiotic detoxification. P450s can operate on a remarkably large range of substrates from the very small to the very large, yet the overall P450 three-dimensional structure is conserved. Given this conservation of structure, it is generally assumed that the basic catalytic mechanism is conserved. In nearly all P450s, the O2 O-O bond must be cleaved heterolytically enabling one oxygen atom, the distal oxygen, to depart as water and leave behind a heme iron-linked O atom as the powerful oxidant that is used to activate the nearby substrate. For this process to proceed efficiently, externally supplied electrons and protons are required. Two protons must be added to the departing O atom while an electron is transferred from a redox partner that typically contains either a Fe2S2 or FMN redox center. The paradigm P450 used to unravel the details of these mechanisms has been the bacterial CYP101A1 or P450cam. P450cam is specific for its own Fe2S2 redox partner, putidaredoxin or Pdx, and it has long been postulated that Pdx plays an effector/allosteric role by possibly switching P450cam to an active conformation. Crystal structures, spectroscopic data, and direct binding experiments of the P450cam-Pdx complex provide some answers. Pdx shifts the conformation of P450cam to a more open state, a transition that is postulated to trigger the proton relay network required for O2 activation. An essential part of this proton relay network is a highly conserved Asp (sometimes Glu) that is known to be critical for activity in a number of P450s. How this Asp and proton delivery networks are connected to redox partner binding is quite simple. In the closed state, this Asp is tied down by salt bridges, but these salt bridges are ruptured when Pdx binds, leaving the Asp free to serve its role in proton transfer. An alternative hypothesis suggests that a specific proton relay network is not really necessary. In this scenario, the Asp plays a structural role in the open/close transition and merely opening the active site access channel is sufficient to enable solvent protons in for O2 protonation. Experiments designed to test these various hypotheses have revealed some surprises in both P450cam and other bacterial P450s. Molecular dynamics and crystallography show that P450cam can undergo rather significant conformational gymnastics that result in a large restructuring of the active site requiring multiple cis/trans proline isomerizations. It also has been found that X-ray driven substrate hydroxylation is a useful tool for better understanding the role that the essential Asp and surrounding residues play in catalysis. Here we summarize these recent results which provide a much more dynamic picture of P450 catalysis.
- Published
- 2022
48. Dynamic assembly of the mRNA m6A methyltransferase complex is regulated by METTL3 phase separation.
- Author
-
Han, Dasol, Longhini, Andrew P, Zhang, Xuemei, Hoang, Vivian, Wilson, Maxwell Z, and Kosik, Kenneth S
- Subjects
Cell Line ,Tumor ,Hela Cells ,Cell Nucleus ,Humans ,Neoplasms ,Multiprotein Complexes ,Methyltransferases ,S-Adenosylmethionine ,Luminescent Proteins ,RNA ,Messenger ,Microscopy ,Confocal ,Catalytic Domain ,Protein Binding ,Mutation ,Cryptochromes ,HEK293 Cells ,HeLa Cells ,Genetics ,Generic health relevance ,Biological Sciences ,Agricultural and Veterinary Sciences ,Medical and Health Sciences ,Developmental Biology - Abstract
m6A methylation is the most abundant and reversible chemical modification on mRNA with approximately one-fourth of eukaryotic mRNAs harboring at least one m6A-modified base. The recruitment of the mRNA m6A methyltransferase writer complex to phase-separated nuclear speckles is likely to be crucial in its regulation; however, control over the activity of the complex remains unclear. Supported by our observation that a core catalytic subunit of the methyltransferase complex, METTL3, is endogenously colocalized within nuclear speckles as well as in noncolocalized puncta, we tracked the components of the complex with a Cry2-METTL3 fusion construct to disentangle key domains and interactions necessary for the phase separation of METTL3. METTL3 is capable of self-interaction and likely provides the multivalency to drive condensation. Condensates in cells necessarily contain myriad components, each with partition coefficients that establish an entropic barrier that can regulate entry into the condensate. In this regard, we found that, in contrast to the constitutive binding of METTL14 to METTL3 in both the diffuse and the dense phase, WTAP only interacts with METTL3 in dense phase and thereby distinguishes METTL3/METTL14 single complexes in the dilute phase from METTL3/METTL14 multicomponent condensates. Finally, control over METTL3/METTL14 condensation is determined by its small molecule cofactor, S-adenosylmethionine (SAM), which regulates conformations of two gate loops, and some cancer-associated mutations near gate loops can impair METTL3 condensation. Therefore, the link between SAM binding and the control of writer complex phase state suggests that the regulation of its phase state is a potentially critical facet of its functional regulation.
- Published
- 2022
49. Imaging active site chemistry and protonation states: NMR crystallography of the tryptophan synthase α-aminoacrylate intermediate
- Author
-
Holmes, Jacob B, Liu, Viktoriia, Caulkins, Bethany G, Hilario, Eduardo, Ghosh, Rittik K, Drago, Victoria N, Young, Robert P, Romero, Jennifer A, Gill, Adam D, Bogie, Paul M, Paulino, Joana, Wang, Xiaoling, Riviere, Gwladys, Bosken, Yuliana K, Struppe, Jochem, Hassan, Alia, Guidoulianov, Jevgeni, Perrone, Barbara, Mentink-Vigier, Frederic, Chang, Chia-En A, Long, Joanna R, Hooley, Richard J, Mueser, Timothy C, Dunn, Michael F, and Mueller, Leonard J
- Subjects
Biochemistry and Cell Biology ,Inorganic Chemistry ,Chemical Sciences ,Biological Sciences ,Alanine ,Catalysis ,Catalytic Domain ,Crystallography ,X-Ray ,Indoles ,Magnetic Resonance Imaging ,Magnetic Resonance Spectroscopy ,Nuclear Magnetic Resonance ,Biomolecular ,Pyridoxal Phosphate ,Tryptophan Synthase ,NMR-assisted crystallography ,tryptophan synthase ,pyridoxal-5 '-phosphate ,integrated structural biology ,solid-state NMR ,pyridoxal-5′-phosphate - Abstract
NMR-assisted crystallography-the integrated application of solid-state NMR, X-ray crystallography, and first-principles computational chemistry-holds significant promise for mechanistic enzymology: by providing atomic-resolution characterization of stable intermediates in enzyme active sites, including hydrogen atom locations and tautomeric equilibria, NMR crystallography offers insight into both structure and chemical dynamics. Here, this integrated approach is used to characterize the tryptophan synthase α-aminoacrylate intermediate, a defining species for pyridoxal-5'-phosphate-dependent enzymes that catalyze β-elimination and replacement reactions. For this intermediate, NMR-assisted crystallography is able to identify the protonation states of the ionizable sites on the cofactor, substrate, and catalytic side chains as well as the location and orientation of crystallographic waters within the active site. Most notable is the water molecule immediately adjacent to the substrate β-carbon, which serves as a hydrogen bond donor to the ε-amino group of the acid-base catalytic residue βLys87. From this analysis, a detailed three-dimensional picture of structure and reactivity emerges, highlighting the fate of the L-serine hydroxyl leaving group and the reaction pathway back to the preceding transition state. Reaction of the α-aminoacrylate intermediate with benzimidazole, an isostere of the natural substrate indole, shows benzimidazole bound in the active site and poised for, but unable to initiate, the subsequent bond formation step. When modeled into the benzimidazole position, indole is positioned with C3 in contact with the α-aminoacrylate Cβ and aligned for nucleophilic attack. Here, the chemically detailed, three-dimensional structure from NMR-assisted crystallography is key to understanding why benzimidazole does not react, while indole does.
- Published
- 2022
50. Lipoprotein-associated phospholipase A2: A paradigm for allosteric regulation by membranes
- Author
-
Mouchlis, Varnavas D, Hayashi, Daiki, Vasquez, Alexis M, Cao, Jian, McCammon, J Andrew, and Dennis, Edward A
- Subjects
Biochemistry and Cell Biology ,Chemical Sciences ,Biological Sciences ,1-Alkyl-2-acetylglycerophosphocholine Esterase ,Allosteric Regulation ,Binding Sites ,Catalysis ,Catalytic Domain ,Fatty Acids ,Humans ,Hydrolysis ,Lipoproteins ,HDL ,Membranes ,Molecular Dynamics Simulation ,Phospholipids ,Substrate Specificity ,phospholipase ,lipoprotein ,allosterism ,membrane ,lipid - Abstract
Lipoprotein-associated phospholipase A2 (Lp-PLA2) associates with low- and high-density lipoproteins in human plasma and specifically hydrolyzes circulating oxidized phospholipids involved in oxidative stress. The association of this enzyme with the lipoprotein's phospholipid monolayer to access its substrate is the most crucial first step in its catalytic cycle. The current study demonstrates unequivocally that a significant movement of a major helical peptide region occurs upon membrane binding, resulting in a large conformational change upon Lp-PLA2 binding to a phospholipid surface. This allosteric regulation of an enzyme's activity by a large membrane-like interface inducing a conformational change in the catalytic site defines a unique dimension of allosterism. The mechanism by which this enzyme associates with phospholipid interfaces to select and extract a single phospholipid substrate molecule and carry out catalysis is key to understanding its physiological functioning. A lipidomics platform was employed to determine the precise substrate specificity of human recombinant Lp-PLA2 and mutants. This study uniquely elucidates the association mechanism of this enzyme with membranes and its resulting conformational change as well as the extraction and binding of specific oxidized and short acyl-chain phospholipid substrates. Deuterium exchange mass spectrometry coupled with molecular dynamics simulations was used to define the precise specificity of the subsite for the oxidized fatty acid at the sn-2 position of the phospholipid backbone. Despite the existence of several crystal structures of this enzyme cocrystallized with inhibitors, little was understood about Lp-PLA2's specificity toward oxidized phospholipids.
- Published
- 2022
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.